Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Int J Mol Sci ; 22(16)2021 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-34445691

RESUMO

The essential role of G-protein coupled receptors (GPCRs) in tumor growth is recognized, yet a GPCR based drug in cancer is rare. Understanding the molecular path of a tumor driver gene may lead to the design and development of an effective drug. For example, in members of protease-activated receptor (PAR) family (e.g., PAR1 and PAR2), a novel PH-binding motif is allocated as critical for tumor growth. Animal models have indicated the generation of large tumors in the presence of PAR1 or PAR2 oncogenes. These tumors showed effective inhibition when the PH-binding motif was either modified or were inhibited by a specific inhibitor targeted to the PH-binding motif. In the second part of the review we discuss several aspects of some cardinal GPCRs in tumor angiogenesis.


Assuntos
Neoplasias/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores Ativados por Proteinase/metabolismo , Expressão Gênica/genética , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Neoplasias/genética , Neovascularização Patológica/metabolismo , Neovascularização Patológica/fisiopatologia , Domínios de Homologia à Plecstrina/genética , Domínios de Homologia à Plecstrina/fisiologia , Domínios Proteicos/genética , Domínios Proteicos/fisiologia , Receptor PAR-1/metabolismo , Receptor PAR-2/metabolismo , Receptores Acoplados a Proteínas G/fisiologia , Receptores Ativados por Proteinase/genética , Transdução de Sinais/fisiologia
2.
Mol Biol Cell ; 32(14): 1306-1319, 2021 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-33979205

RESUMO

The neuronal dynamin1 functions in the release of synaptic vesicles by orchestrating the process of GTPase-dependent membrane fission. Dynamin1 associates with the plasma membrane-localized phosphatidylinositol-4,5-bisphosphate (PIP2) through the centrally located pleckstrin homology domain (PHD). The PHD is dispensable as fission (in model membranes) can be managed, even when the PHD-PIP2 interaction is replaced by a generic polyhistidine- or polylysine-lipid interaction. However, the absence of the PHD renders a dramatic dampening of the rate of fission. These observations suggest that the PHD-PIP2-containing membrane interaction could have evolved to expedite fission to fulfill the requirement of rapid kinetics of synaptic vesicle recycling. Here, we use a suite of multiscale modeling approaches to explore PHD-membrane interactions. Our results reveal that 1) the binding of PHD to PIP2-containing membranes modulates the lipids toward fission-favoring conformations and softens the membrane, and 2) PHD associates with membrane in multiple orientations using variable loops as pivots. We identify a new loop (VL4), which acts as an auxiliary pivot and modulates the orientation flexibility of PHD on the membrane-a mechanism that we believe may be important for high-fidelity dynamin collar assembly. Together, these insights provide a molecular-level understanding of the catalytic role of PHD in dynamin-mediated membrane fission.


Assuntos
Dinamina I/metabolismo , Domínios de Homologia à Plecstrina/fisiologia , Proteínas Sanguíneas/metabolismo , Proteínas Sanguíneas/fisiologia , Catálise , Membrana Celular/metabolismo , Biologia Computacional/métodos , Dinamina I/química , Dinamina I/fisiologia , Dinaminas/metabolismo , Endocitose/fisiologia , GTP Fosfo-Hidrolases/metabolismo , Guanosina Trifosfato/metabolismo , Humanos , Hidrólise , Membranas/metabolismo , Fosfatidilinositol 4,5-Difosfato/metabolismo , Fosfatidilinositóis/metabolismo , Fosfoproteínas/metabolismo , Fosfoproteínas/fisiologia , Multimerização Proteica , Estrutura Terciária de Proteína , Relação Estrutura-Atividade , Vesículas Sinápticas/fisiologia
3.
Mol Biol Cell ; 31(2): 101-117, 2020 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-31774725

RESUMO

Myosin 1s have critical roles in linking membranes to the actin cytoskeleton via direct binding to acidic lipids. Lipid binding may occur through PIP3/PIP2-specific PH domains or nonspecific ionic interactions involving basic-hydrophobic (BH) sites but the mechanism of myosin 1s distinctive lipid targeting is poorly understood.  Now we show that PH domains occur in all Dictyostelium myosin 1s and that the BH sites of Myo1A, B, C, D, and F are in conserved positions near the ß3/ß4 loops of their PH domains. In spite of these shared lipid-binding sites, we observe significant differences in myosin 1s highly dynamic localizations. All myosin 1s except Myo1A are present in macropinocytic structures but only Myo1B and Myo1C are enriched at the edges of macropinocytic cups and associate with the actin in actin waves.  In contrast, Myo1D, E, and F are enclosed by the actin wave.  Mutations of BH sites affect localization of all Dictyostelium myosin 1s. Notably, mutation of the BH site located within the PH domains of PIP3-specific Myo1D and Myo1F completely eradicates membrane binding. Thus, BH sites are important determinants of motor targeting and may have a similar role in the localization of other myosin 1s.


Assuntos
Dictyostelium/metabolismo , Miosina Tipo I/metabolismo , Domínios de Homologia à Plecstrina/fisiologia , Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Sítios de Ligação , Membrana Celular/metabolismo , Interações Hidrofóbicas e Hidrofílicas , Miosina Tipo I/genética , Miosina Tipo I/ultraestrutura , Miosinas/metabolismo , Domínios Proteicos/fisiologia , Transporte Proteico , Proteínas de Protozoários/metabolismo
4.
Cells ; 8(5)2019 05 17.
Artigo em Inglês | MEDLINE | ID: mdl-31108912

RESUMO

Cytokinesis D is known as the midwife mechanism in which neighboring cells facilitate cell division by crossing the cleavage furrow of dividing cells. Cytokinesis D is thought to be mediated by chemotaxis, where midwife cells migrate toward dividing cells by sensing an unknown chemoattractant secreted from the cleavage furrow. In this study, to validate this chemotaxis model, we aspirated the fluid from the vicinity of the cleavage furrow of a dividing Dictyostelium cell and discharged it onto a neighboring cell using a microcapillary. However, the neighboring cells did not show any chemotaxis toward the fluid. In addition, the cells did not manifest an increase in the levels of intracellular Ca2+, cAMP, or cGMP, which are expected to rise in chemotaxing cells. From several lines of our experiments, including these findings, we concluded that chemotaxis does not contribute to cytokinesis D. As an alternative, we propose a cortical-flow model, where a migrating cell attaches to a dividing cell by chance and is guided toward the furrow by the cortical flow on the dividing cell, and then physically assists the separation of the daughter cells.


Assuntos
Quimiotaxia/fisiologia , Citocinese/fisiologia , Dictyostelium/citologia , Dictyostelium/fisiologia , Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Cálcio/metabolismo , Movimento Celular , Rastreamento de Células/métodos , Células Cultivadas , Fatores Quimiotáticos/metabolismo , AMP Cíclico/metabolismo , GMP Cíclico/metabolismo , Citoplasma/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Interfase/fisiologia , Microscopia de Contraste de Fase , Mitose/fisiologia , Modelos Biológicos , Domínios de Homologia à Plecstrina/fisiologia
5.
Nat Chem Biol ; 15(4): 358-366, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30742123

RESUMO

Peripheral membrane proteins orchestrate many physiological and pathological processes, making regulation of their activities by small molecules highly desirable. However, they are often refractory to classical competitive inhibition. Here, we demonstrate that potent and selective inhibition of peripheral membrane proteins can be achieved by small molecules that target protein-membrane interactions by a noncompetitive mechanism. We show that the small molecule Bragsin inhibits BRAG2-mediated Arf GTPase activation in vitro in a manner that requires a membrane. In cells, Bragsin affects the trans-Golgi network in a BRAG2- and Arf-dependent manner. The crystal structure of the BRAG2-Bragsin complex and structure-activity relationship analysis reveal that Bragsin binds at the interface between the PH domain of BRAG2 and the lipid bilayer to render BRAG2 unable to activate lipidated Arf. Finally, Bragsin affects tumorsphere formation in breast cancer cell lines. Bragsin thus pioneers a novel class of drugs that function by altering protein-membrane interactions without disruption.


Assuntos
Fator 1 de Ribosilação do ADP/fisiologia , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Fatores de Troca do Nucleotídeo Guanina/fisiologia , Fator 1 de Ribosilação do ADP/metabolismo , Linhagem Celular Tumoral , GTP Fosfo-Hidrolases , Proteínas Ativadoras de GTPase , Fatores de Troca do Nucleotídeo Guanina/antagonistas & inibidores , Células HeLa , Humanos , Bicamadas Lipídicas , Glicoproteínas de Membrana/metabolismo , Nucleotídeos , Domínios de Homologia à Plecstrina/fisiologia , Ligação Proteica , Transdução de Sinais , Relação Estrutura-Atividade , Sulfotransferases/metabolismo
6.
J Microbiol Biotechnol ; 28(8): 1401-1411, 2018 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-30301316

RESUMO

The serine-threonine kinase AKT plays a pivotal role in tumor progression and is frequently overactivated in cancer cells; this protein is therefore a critical therapeutic target for cancer intervention. We aimed to identify small molecule inhibitors of the pleckstrin homology (PH) domain of AKT to disrupt binding of phosphatidylinositol-3,4,5-trisphosphate (PIP3), thereby downregulating AKT activity. Liposome pulldown assays coupled with fluorescence spectrometry were used to screen flavonoids for inhibition of the AKT PH-PIP3 interaction. Western blotting was used to determine the effects of the inhibitors on AKT activation in cancer cells, and in silico docking was used for structural analysis and optimization of inhibitor structure. Several flavonoids showing up to 50% inhibition of the AKT PH-PIP3 interaction decreased the level of AKT activation at the cellular level. In addition, the modified flavonoid showed increased inhibitory effects and the approach would be applied to develop anticancer drug candidates. In this study, we provide a rationale for targeting the lipid-binding domain of AKT, rather than the catalytic kinase domain, in anticancer drug development.


Assuntos
Antineoplásicos/farmacologia , Flavonoides/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Domínios de Homologia à Plecstrina/fisiologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Antineoplásicos/química , Antineoplásicos/classificação , Antineoplásicos/metabolismo , Sítios de Ligação , Linhagem Celular Tumoral , Descoberta de Drogas , Flavonoides/química , Flavonoides/farmacologia , Humanos , Lipossomos/química , Lipossomos/metabolismo , Modelos Moleculares , Simulação de Acoplamento Molecular , Estrutura Molecular , Fosfatos de Fosfatidilinositol/química , Domínios de Homologia à Plecstrina/genética , Ligação Proteica/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/química , Relação Quantitativa Estrutura-Atividade , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo
7.
Sci Rep ; 8(1): 12910, 2018 08 27.
Artigo em Inglês | MEDLINE | ID: mdl-30150713

RESUMO

Targeting proteins to regions where they are required is essential for proper development of organisms. For achievement of this, subcellular mRNA localization is one of the critical mechanisms. Subcellular mRNA localization is an evolutionarily conserved phenomenon from E. coli to human and contributes to limiting the regions at which its products function and efficiently supplies substrates for protein translation. During early Drosophila embryogenesis, while 71% of the 3370 mRNAs analyzed have shown prominent subcellular localization, the underlying molecular mechanisms have not been elucidated. Here, we reveal that anillin mRNA, one of the localized mRNAs in early Drosophila embryo, localizes to the tip of the pseudo-cleavage furrow in the Drosophila syncytial blastoderm using in situ hybridization combined with immunohistochemistry. Localization analyses with transgenic fly lines carrying a series of deletion mRNAs indicate that this localization is dependent on its own nascent polypeptides including the actin binding domain (ABD). In addition to the mRNA localization, it is revealed that the pleckstrin homology (PH) domain of Anillin protein is also required for its proper localization. Thus, we indicate that the precise localization of Anillin protein is tightly regulated by the ABD on the nascent polypeptide and PH domain in the Drosophila syncytial blastoderm.


Assuntos
Proteínas Contráteis/metabolismo , Proteínas de Drosophila/metabolismo , Proteínas dos Microfilamentos/metabolismo , Peptídeos/metabolismo , Animais , Blastoderma/metabolismo , Ciclo Celular/genética , Ciclo Celular/fisiologia , Proteínas Contráteis/genética , Drosophila , Proteínas de Drosophila/genética , Proteínas dos Microfilamentos/genética , Peptídeos/genética , Domínios de Homologia à Plecstrina/genética , Domínios de Homologia à Plecstrina/fisiologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
8.
Proc Natl Acad Sci U S A ; 115(25): E5766-E5775, 2018 06 19.
Artigo em Inglês | MEDLINE | ID: mdl-29866840

RESUMO

The identification and targeting of key molecular drivers of melanoma and breast and lung cancer have substantially improved their therapy. However, subtypes of each of these three common, lethal solid tumors lack identified molecular drivers, and are thus not amenable to targeted therapies. Here we show that pleckstrin homology domain-interacting protein (PHIP) promotes the progression of these "driver-negative" tumors. Suppression of PHIP expression significantly inhibited both tumor cell proliferation and invasion, coordinately suppressing phosphorylated AKT, cyclin D1, and talin1 expression in all three tumor types. Furthermore, PHIP's targetable bromodomain is functional, as it specifically binds the histone modification H4K91ac. Analysis of TCGA profiling efforts revealed PHIP overexpression in triple-negative and basal-like breast cancer, as well as in the bronchioid subtype of nonsmall cell lung cancer. These results identify a role for PHIP in the progression of melanoma and breast and lung cancer subtypes lacking identified targeted therapies. The use of selective, anti-PHIP bromodomain inhibitors may thus yield a broad-based, molecularly targeted therapy against currently nontargetable tumors.


Assuntos
Mama/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Neoplasias Pulmonares/metabolismo , Melanoma/metabolismo , Domínios de Homologia à Plecstrina/fisiologia , Neoplasias de Mama Triplo Negativas/metabolismo , Animais , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/fisiologia , Ciclina D1/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica/fisiologia , Humanos , Proteínas Proto-Oncogênicas c-akt/metabolismo
9.
Biochem Biophys Res Commun ; 496(4): 1088-1094, 2018 02 19.
Artigo em Inglês | MEDLINE | ID: mdl-29409900

RESUMO

Oxysterol-binding protein-related proteins are implicated in the sensing and transporting lipids at the membrane contact sites. One of the members of the mammalian ORP family, ORP8, is thought to transport lipids through directly tethering both ER and PM membranes. Targeting to PM is thought to be mediated by N-terminal pleckstrin homology domain via binding to phosphoinositides. Sequence alignments and NMR structural determination revealed that the PH domain of ORP8 is atypical and contains an insertion of 20 amino acids in an unstructured loop region that may potentially block interactions with ligands. Using standard lipid-protein overlay assays or liposomal binding assays we could not detect binding of a recombinant version of the PH domain. Examination of a series of deletion constructs demonstrated that both the N-terminal polybasic region and the PH domain are required for proper targeting of the short splice variant ORP8S to the PM-ER contact site in Chinese hamster ovary cells.


Assuntos
Membrana Celular/química , Membrana Celular/metabolismo , Retículo Endoplasmático/química , Retículo Endoplasmático/metabolismo , Domínios de Homologia à Plecstrina/fisiologia , Receptores de Esteroides/química , Receptores de Esteroides/metabolismo , Animais , Sítios de Ligação , Células CHO , Membrana Celular/ultraestrutura , Cricetulus , Retículo Endoplasmático/ultraestrutura , Bicamadas Lipídicas/química , Ligação Proteica
10.
Proc Natl Acad Sci U S A ; 114(45): 11926-11931, 2017 11 07.
Artigo em Inglês | MEDLINE | ID: mdl-29078297

RESUMO

Insulin resistance, a key etiological factor in metabolic syndrome, is closely linked to ectopic lipid accumulation and increased intracellular Ca2+ concentrations in muscle and liver. However, the mechanism by which dysregulated intracellular Ca2+ homeostasis causes insulin resistance remains elusive. Here, we show that increased intracellular Ca2+ acts as a negative regulator of insulin signaling. Chronic intracellular Ca2+ overload in hepatocytes during obesity and hyperlipidemia attenuates the phosphorylation of protein kinase B (Akt) and its key downstream signaling molecules by inhibiting membrane localization of pleckstrin homology (PH) domains. Pharmacological approaches showed that elevated intracellular Ca2+ inhibits insulin-stimulated Akt phosphorylation and abrogates membrane localization of various PH domain proteins such as phospholipase Cδ and insulin receptor substrate 1, suggesting a common mechanism inhibiting the membrane targeting of PH domains. PH domain-lipid overlay assays confirmed that Ca2+ abolishes the binding of various PH domains to phosphoinositides (PIPs) with two adjacent phosphate groups, such as PI(3,4)P2, PI(4,5)P2, and PI(3,4,5)P3 Finally, thermodynamic analysis of the binding interaction showed that Ca2+-mediated inhibition of targeting PH domains to the membrane resulted from the tight binding of Ca2+ rather than PH domains to PIPs forming Ca2+-PIPs. Thus, Ca2+-PIPs prevent the recognition of PIPs by PH domains, potentially due to electrostatic repulsion between positively charged side chains in PH domains and the Ca2+-PIPs. Our findings provide a mechanistic link between intracellular Ca2+ dysregulation and Akt inactivation in insulin resistance.


Assuntos
Cálcio/metabolismo , Membrana Celular/metabolismo , Resistência à Insulina/fisiologia , Fosfatidilinositóis/metabolismo , Domínios de Homologia à Plecstrina/fisiologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Animais , Dieta Hiperlipídica , Intolerância à Glucose/patologia , Hiperinsulinismo/patologia , Insulina/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Obesidade/patologia , Fosfolipase C delta/metabolismo , Fosforilação , Ligação Proteica
11.
Biochim Biophys Acta Proteins Proteom ; 1865(10): 1274-1286, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28698152

RESUMO

BACKGROUND: Three NPXY motifs and one FERM domain in CCM1 makes it a versatile scaffold protein for tethering the signaling components together within the CCM signaling complex (CSC). The cellular role of CCM1 protein remains inadequately expounded. Both phosphotyrosine binding (PTB) and pleckstrin homology (PH) domains were recognized as structurally related but functionally distinct domains. METHODS: By utilizing molecular cloning, protein binding assays and RT-qPCR to identify novel cellular partners of CCM1 and its cellular expression patterns; by screening candidate PTB/PH proteins and subsequently structurally simulation in combining with current X-ray crystallography and NMR data to defined the essential structure of PTB/PH domain for NPXY-binding and the relationship among PTB, PH and FERM domain(s). RESULTS: We identified a group of 28 novel cellular partners of CCM1, all of which contain either PTB or PH domain(s), and developed a novel classification system for these PTB/PH proteins based on their relationship with different NPXY motifs of CCM1. Our results demonstrated that CCM1 has a wide spectrum of binding to different PTB/PH proteins and perpetuates their specificity to interact with certain PTB/PH domains through selective combination of three NPXY motifs. We also demonstrated that CCM1 can be assembled into oligomers through intermolecular interaction between its F3 lobe in FERM domain and one of the three NPXY motifs. Despite being embedded in FERM domain as F3 lobe, F3 module acts as a fully functional PH domain to interact with NPXY motif. The most salient feature of the study was that both PTB and PH domains are structurally and functionally comparable, suggesting that PTB domain is likely evolved from PH domain with polymorphic structural additions at its N-terminus. CONCLUSIONS: A new ß1A-strand of the PTB domain was discovered and new minimum structural requirement of PTB/PH domain for NPXY motif-binding was determined. Based on our data, a novel theory of structure, function and relationship of PTB, PH and FERM domains has been proposed, which extends the importance of the NPXY-PTB/PH interaction on the CSC signaling and/or other cell receptors with great potential pointing to new therapeutic strategies. GENERAL SIGNIFICANCE: The study provides new insight into the structural characteristics of PTB/PH domains, essential structural elements of PTB/PH domain required for NPXY motif-binding, and function and relationship among PTB, PH and FERM domains.


Assuntos
Proteínas Sanguíneas/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Fosfoproteínas/metabolismo , Fosfotirosina/metabolismo , Domínios de Homologia à Plecstrina/fisiologia , Domínios Proteicos/fisiologia , Motivos de Aminoácidos , Sítios de Ligação , Ligação Proteica
12.
Neurobiol Learn Mem ; 138: 31-38, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-27344941

RESUMO

ADP-ribosylation factors (ARFs) are small guanosine triphosphatases of the Ras superfamily involved in membrane trafficking and regulation of the actin cytoskeleton. Aplysia Sec7 protein (ApSec7), a guanine nucleotide exchange factor for ARF1 and ARF6, induces neurite outgrowth and plays a key role in 5-hydroxyltryptamine-induced neurite growth and synaptic facilitation in Aplysia sensory-motor synapses. However, the specific role of ARF6 signaling on neurite outgrowth in Aplysia neurons has not been examined. In the present study, we cloned Aplysia ARF6 (ApARF6) and revealed that an overexpression of enhanced green fluorescent protein (EGFP)-fused constitutively active ApARF6 (ApARF6-Q67L-EGFP) could induce neurite outgrowth in Aplysia sensory neurons. Further, we observed that ApARF6-induced neurite outgrowth was inhibited by the co-expression of a Sec7 activity-deficient mutant of ApSec7 (ApSec7-E159K). The pleckstrin homology domain of ApSec7 may bind to active ApARF6 at the plasma membrane and prevent active ApARF6-induced functions, including intracellular vacuole formation in HEK293T cells. The results of the present study suggest that activation of ARF6 signaling could induce neurite outgrowth in Aplysia neurons and may be involved in downstream signaling of ApSec7-induced neurite outgrowth in Aplysia neurons.


Assuntos
Fatores de Ribosilação do ADP/metabolismo , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Neuritos/metabolismo , Crescimento Neuronal/fisiologia , Domínios de Homologia à Plecstrina/fisiologia , Células Receptoras Sensoriais/metabolismo , Animais , Aplysia , Células HEK293 , Humanos , Transdução de Sinais/fisiologia , Sinapses/metabolismo , Regulação para Cima
13.
Mol Biol Cell ; 28(1): 152-160, 2017 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-28035046

RESUMO

Classical dynamins bind the plasma membrane-localized phosphatidylinositol-4,5-bisphosphate using the pleckstrin-homology domain (PHD) and engage in rapid membrane fission during synaptic vesicle recycling. This domain is conspicuously absent among extant bacterial and mitochondrial dynamins, however, where loop regions manage membrane recruitment. Inspired by the core design of bacterial and mitochondrial dynamins, we reengineered the classical dynamin by replacing its PHD with a polyhistidine or polylysine linker. Remarkably, when recruited via chelator or anionic lipids, respectively, the reengineered dynamin displayed the capacity to constrict and sever membrane tubes. However, when analyzed at single-event resolution, the tube-severing process displayed long-lived, highly constricted prefission intermediates that contributed to 10-fold reduction in bulk rates of membrane fission. Our results indicate that the PHD acts as a catalyst in dynamin-induced membrane fission and rationalize its adoption to meet the physiologic requirement of a fast-acting membrane fission apparatus.


Assuntos
Dinaminas/metabolismo , Dinaminas/ultraestrutura , Dinâmica Mitocondrial/fisiologia , Membrana Celular/metabolismo , Constrição , Dinaminas/genética , Endocitose/fisiologia , GTP Fosfo-Hidrolases/metabolismo , Guanosina Trifosfato/metabolismo , Humanos , Membranas/metabolismo , Membranas/fisiologia , Mitocôndrias/metabolismo , Dinâmica Mitocondrial/genética , Fosfatidilinositol 4,5-Difosfato/metabolismo , Fosfatidilinositóis/metabolismo , Domínios de Homologia à Plecstrina/genética , Domínios de Homologia à Plecstrina/fisiologia , Domínios Proteicos , Estrutura Terciária de Proteína
14.
PLoS One ; 11(12): e0168806, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-28036396

RESUMO

Protein kinase B-ß (PKBß/Akt2) is a serine/threonine-specific protein kinase that has emerged as one of the most important regulators of cell growth, differentiation, and division. Upregulation of Akt2 in various human carcinomas, including ovarian, breast, and pancreatic, is a well-known tumorigenesis phenomenon. Early on, the concept of the simultaneous administration of anticancer drugs with inhibitors of Akt2 was advocated to overcome cell proliferation in the chemotherapeutic treatment of cancer. However, clinical studies have not lived up to the high expectations, and several phase II and phase III clinical studies have been terminated prematurely because of severe side effects related to the non-selective isomeric inhibition of Akt2. The notion that the sequence identity of pleckstrin homology (PH) domains within Akt-isoforms is less than 30% might indicate the possibility of the development of selective antagonists against the Akt2 PH domain. Therefore, in this study, various in silico tools were utilized to explore the hypothesis that quinoline-type inhibitors bind in the Akt2 PH domain. A Grid-Independent Molecular Descriptor (GRIND) analysis indicated that two hydrogen bond acceptors, two hydrogen bond donors and one hydrophobic feature at a certain distance from each other were important for the selective inhibition of Akt2. Our docking results delineated the importance of Lys30 as an anchor point for mapping the distances of important amino acid residues in the binding pocket, including Lys14, Glu17, Arg25, Asn53, Asn54 and Arg86. The binding regions identified complement the GRIND-based pharmacophoric features.


Assuntos
Antineoplásicos/farmacologia , Domínios de Homologia à Plecstrina/fisiologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Quinolinas/farmacologia , Aminoácidos/metabolismo , Sítios de Ligação , Humanos , Ligação de Hidrogênio , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Isoformas de Proteínas/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Relação Quantitativa Estrutura-Atividade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...